Language selection

Search

Patent 2224257 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2224257
(54) English Title: RECOMBINANT RACCOON POX VIRUSES AND THEIR USE AS AN EFFECTIVE VACCINE AGAINST FELINE IMMUNODEFICIENCY VIRUS INFECTION
(54) French Title: POXVIRUS RECOMBINES DU RATON LAVEUR ET LEUR UTILISATION EN TANT QUE VACCIN EFFICACE CONTRE L'INFECTION PAR LE VIRUS DE L'IMMUNODEFICIENCE FELINE
Status: Deemed expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 7/01 (2006.01)
  • A61K 39/21 (2006.01)
  • A61K 39/275 (2006.01)
  • A61K 39/295 (2006.01)
  • A61P 31/12 (2006.01)
  • C07K 14/155 (2006.01)
  • C12N 15/863 (2006.01)
  • A61K 39/00 (2006.01)
(72) Inventors :
  • WASMOEN, TERRI (United States of America)
  • CHU, HSIEN-JUE (United States of America)
  • CHAVEZ, LLOYD GEORGE, JR. (United States of America)
(73) Owners :
  • WYETH (United States of America)
(71) Applicants :
  • AMERICAN HOME PRODUCTS CORPORATION (United States of America)
(74) Agent: RIDOUT & MAYBEE LLP
(74) Associate agent:
(45) Issued: 2005-08-16
(86) PCT Filing Date: 1996-06-03
(87) Open to Public Inspection: 1996-12-19
Examination requested: 2001-05-30
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US1996/008508
(87) International Publication Number: WO1996/040268
(85) National Entry: 1997-12-05

(30) Application Priority Data:
Application No. Country/Territory Date
08/482,090 United States of America 1995-06-07

Abstracts

English Abstract



The present invention provides recombinant raccoon poxviruses (RRPVs) useful
in vaccines for the prophylaxis of disease caused by
feline immunodeficiency virus (FIV). RRPVs according to the invention have at
least one internal gene comprising a DNA sequence that
encodes FIV gag protein (gag), FIV enveloppe protein (env), a polypeptide
consisting of amino acids 1-735 of FIV env, or immunogenic
fragments of any of the foregoing. The vaccines that comprise one or more of
the FIV-expressing recombinant raccoon poxviruses described
above may also comprise a pharmaceutically acceptable carrier or diluent and a
pharmaceutically acceptable adjuvant. The invention also
provides methods for preventing or lessening disease caused by FIV, which is
carried out by administering to a feline in need of such
treatment the vaccines described above.


French Abstract

L'invention concerne des poxyvirus recombinés du raton laveur (RRPVs) utiles dans les vaccins pour la prophylaxie de maladies causées par le virus de l'immunodéficience féline (VIF). Les RRPVs, selon cette invention, possèdent au moins un gène interne comprenant une séquence d'ADN qui code pour la protéine gag du VIF (gag), la protéine enveloppe du VIF (env), un polypeptide formé des acides aminés 1-735 d'env du VIF, ou des fragments immunogènes de l'une quelconque des substances précitées. Les vaccins qui comprennent un ou plusieurs poxvirus recombinés du raton laveur exprimant le VIF décrits plus haut peuvent également comprendre un vecteur ou un diluant pharmaceutiquement acceptable ainsi qu'un adjuvant pharmaceutiquement acceptable. L'invention décrit également des procédés pour la prévention ou la réduction de maladies causées par le VIF, consistant à administrer à un félin nécessitant ce type de traitement les vaccins précités.

Claims

Note: Claims are shown in the official language in which they were submitted.



24

CLAIMS:

1. A recombinant raccoon poxvirus having at least one internal gene
comprising a DNA sequence encoding the envelope protein of Feline
Immunodeficiency
Vints (FIV) or immunogenic fragments therefrom.

2. The recombinant raccoon poxvirus of claim 1 wherein said internal
gene encodes the FIV envelope protein having the amino acid sequence as set
out in
Figure 3 or immunogenic fragments therefrom.

3. The recombinant raccoon poxvirus of claim 2 wherein said internal
gene encodes amino acids 1-735 of the FIV envelope protein.

4. A vaccine comprising:
a recombinant raccoon poxvirus having at least one internal gene
comprising a DNA sequence encoding envelope protein of Feline Immunodeficiency
Virus
(FIV) or immunogenic fragments therefrom, and
a pharmaceutically acceptable carrier or diluent.

5. The vaccine of claim 4 further comprising a pharmaceutically
acceptable adjuvant.

6. The vaccine of claim 4. wherein said internal gene encodes the FIV
envelope protein having the amino acid sequence as set out in Figure 3 or
immunogenic
fragments therefrom.

7. The vaccine of claim 4 wherein said internal gene encodes amino
acids 1-735 of the FIV envelope protein.

8. The vaccine of claim 4 further comprising immunogens derived
from viruses selected from the group consisting of feline leukemia virus,
feline



25

panleucopenia virus, feline rhinotracheitis virus, feline calicivirus, feline
infectious
peritonitis virus, feline herpesvirus, feline enteric coronavirus, or mixtures
thereof.

9. The vaccine of claim 4 further comprising inactivated or attenuated
feline Chlamydia psittaci, Microsporum canis, or mixtures thereof.

10. A recombinant raccoon poxvirus having at least one internal gene
comprising a DNA sequence encoding the gag protein of Feline Immunodeficiency
Virus
(FIV) or immunogenic fragments therefrom.

11. The recombinant raccoon poxvirus of claim 10 wherein said internal
gene encodes the FIV gag protein having the amino acid sequence as set out in
Figure 5 or
immunogenic fragments therefrom.

12. A vaccine comprising:
a recombinant raccoon poxvirus having at least one internal gene
comprising a DNA sequence encoding gag protein of Feline Immunodeficiency
Virus
(FIV) or immunogenic fragments therefrom, and
a pharmaceutically acceptable carrier or diluent.

13. The vaccine of claim 12 further comprising a pharmaceutically
acceptable adjuvant.

14. The vaccine of claim 12 wherein said internal gene encodes the FIV
gag protein having the amino acid sequence as set out in Figure 5 or
immunogenic
fragments therefrom.

15. The vaccine of claim 12 further comprising immunogens derived
from viruses selected from the group consisting of feline leukemia virus,
feline
panleucopenia virus, feline rhinotracheitis virus, feline calicivirus, feline
infectious
peritonitis virus, feline herpesvirus, feline enteric coronavirus, or mixtures
thereof.



-26-

16. The vaccine of claim 12 further comprising
inactivated or attenuated feline Chlamydia psittaci,
Microsporum canis, or mixtures thereof.

17. A vaccine comprising:
a first recombinant raccoon poxvirus having at
least one internal gene comprising a DNA sequence encoding
a member selected from the group consisting of the gag and
envelope proteins of Feline Immunodeficiency Virus (FIV) or
immunogenic fragments therefrom;
a second recombinant raccoon poxvirus having at
least one internal gene comprising a DNA sequence encoding
a member selected form the group consisting of the gag and
envelope proteins of Feline Immunodeficiency Virus (FIV) or
immunogenic fragments therefrom; and
a pharmaceutically acceptable carrier or diluent.

18. The vaccine of claim 17 further comprising a
pharmaceutically acceptable adjuvant.

19. Use of vaccine comprising a recombinant raccoon
poxvirus having at least one internal gene comprising a DNA
sequence encoding the envelope protein of Feline
Immunodeficiency Virus (FIV) or immunogenic fragments
therefrom, for preventing or lessening disease caused by
Feline Immunodeficiency Virus (FIV).

20. The use ref claim 19, wherein said internal gene
encodes the FIV envelope protein having the amino acid
sequence as set out in Figure 3 or immunogenic fragments
thereof.

21. Use of a vaccine comprising a recombinant raccoon
poxvirus having at least one internal gene comprising a DNA



-27-

sequence encoding the gag protein of Feline
Immunodeficiency Virus (FIV) or immunogenic fragments
therefrom, for preventing or lessening disease caused by
Feline Immunodeficiency Virus (FIV).

22. The use of claim 21 wherein said internal gene
encodes the FIV gag protein having the amino acid sequence
as set out in Figure 5 or immunogenic fragments thereof.

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
RECOMBINANT RACCOON POX VIRUSES AND THEIR USE
AS AN EFFECTIVE VACCINE AGAINST FELINE
IIVVIMUNODEFICIENCY VIRUS INFECTION
The present invention pertains to the prophylaxis of disease caused by
feline immunodeficiency virus (FTV), using as vaccines recombinant raccoon
poxviruses
(RRPVs) expressing the gag and envelope proteins of FIV.
~Al.lilxKC)U1V1~ 1' 1 H. INVFN'j'InN
Feline immunodeficiency virus (FIB infection is a significant health
problem for domestic cats around the world. As in its human counterpart,
infection with
FIV causes a progressive disruption in immune function. In the acute phase of
infection,
the virus causes transient illness associated with symptoms such as
lymphadenopathy,
pyrexia, and neutropenia. Subsequently, an infected animal enters an
asymptomatic phase
of 1-2 years before clinical manifestations of immune deficiency become
apparent, after
which the mean survival time is usually less than one year.
FIV is a typical retrovirus that contains a single-stranded polyadenylated
RNA genome, internal structural proteins derived from the gag gene product,
and a lipid
envelope containing membrane proteins derived from the env gene product
(Bendinelli et
al., CIin.Microbiol.Rev. $:87, 1995). The gag gene is translated into a
primary product of
about 50 kDa that is subsequently cleaved by a viral protease into the matrix,
capsid, and
nucleocapsid proteins. The env gene yields a primary translation product of 75-
80 lcDa
(unglycosylated molecular weight); in infected cells, the precursor has an
apparent


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
2 -
molecular weight of. 145-150 kDa due to N-linked glycosylation. The env
precursor is
cleaved in the Golgi apparatus into the SU and TM proteins (also designated
gp95 and
gp40, respectively).
Most vaccines against FIV have failed to induce protective immunity.
Ineffective vaccines have involved inactivated whole virus, fixed infected
cells,
recombinant CA and SU proteins, and a synthetic peptide corresponding to the
V3 region
of SU. In some cases, the vaccine actually enhanced infection after challenge.
In one
system, vaccination with paraformaldehyde-fixed virus or infected cells
resulted in
protective immunity (Yamamoto et al., J. Virol. ø2:601, 1993), but application
of this
approach by others was unsuccessful (Hosie et al., in Abstracts of the
International
Symposium on Feline Retrovirus Research, 1993, page 50).
Thus, there is a need in the art for an effective vaccine against FIV that
utilizes the gag or env proteins, or fragments therefrom, as immunogens.
5U ARy O ~ . nWFNT1n11T
The present invention pertains to the prevention or lessening of disease in
cats caused by Feline Immunodeficiency Virus (FIV). Prevention or lessening of
disease
is understood to mean the amelioration of any symptoms, including immune
system
disruptions, that result from FIV infection.
The invention provides recombinant raccoon poxviruses having at least one
internal gene comprising a DNA sequence that encodes FIV gag protein (gag),
FIV
envelope protein (env), a polypeptide consisting of amino acids 1-735 of FIV
env, or
immunogenic fragments of any of the foregoing. By immunogenic fragment is
meant any
portion of the coding sequence of FIV gag or env polypeptides that induces a
beneficial
immune response in cats.
In another aspect, the invention encompasses vaccines that comprise one or
more of the FIV~xpressing recombinant raccoon poxviruses described above, with
a
pharmaceutically acceptable carrier or diluent and a pharmaceutically
acceptable adjuvant. '
In yet another aspect, the invention provides methods for preventing or
lessening disease caused by FIV, which is carried out by administering to a
feline in need
of such treatment the vaccines described above.


CA 02224257 2003-02-19
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. is a3 graphic: il.l:zstration of the cloning
strategy for the c-nvelope gene of FIV.
Figure % is a diagrammatic: representation of the
structure of_ the u:eccmbinant FIV env gene in pSL-EnvAl3C.
Figure 3 shov~r~s th~~ DNA [SEQ. I..7. N0. 14] and protein
[SEQ. I.D. NO. 12sequence of the env genE~ of FIV.
Figure 4 is .::c graphic i?.l~.xstrat.aon of: the pSL-WGag
plasmicl.
Figure ~ sho~rs the DNA (SEQ. I.D, NO. 13] and protein
[SEQ. I.D. NO. 1.1~ sequence of the gag gene oi= FIV.
Figure e7 is ~c graphic: .ilLustrat:v.~on o:r the cloning
strategy for const::ructi_on of ttue raccoon poxvirus transfer
olasmid pSCl7..-FIV gag .
Figure '~ is ~~ graphic illustration of- the cloning
strategy for const:ructi.on of the rac~.-:oon poxvirus transfer
plasmicl ~>SC1:1..-FIV E:,rm .
Fi.gurc: 8 is a~ graphic i_Llustration c>f the cloning
strategy for const:ructi_on of tine raccoon poxvirus transfer
plasmicl y~SC1:1.-FIV E.',nvAB.
Figure 9 is ~:c tabl.e illustrating the detection o:E
viremia. anc~ C:D4:CI;~B r.~t:i_os .in ,~~accinat.ed and unvaccin<~ted
eats after F:IV ch~cl.lenqc:.
Figure 10 is a t,ai;~.te il.Lu:~tratind the preventable
fraction for viren,i.a arcd CD4 : CIO8 rat io changes in
vaccinated and unvaccinated cats fol:Lowind FIV challenge.
Figure :l.l is a t,~ble i.llu~trating the clinical scores
of vaccinated and unva<::cinate>d cats after challenge w=ith
Toxoplas.ma g~andii .


CA 02224257 2003-02-19
_g~_
DETAILED DESCRIPTION OF THE INVENTION
With regard tc~ the patents, patent a~:~plications, and
references cited toei:ein, in the case of in~:onsistencies,
the prE:sent disclc:~sure, _incl;zding definiti~ms, will
cont rol. .
The vaccine c;f the present: invent=.ion rnay be prep<~red
by creating recomx~inant~ rac:cc>ori poxviruses (RRPVs)
containing a gene encoding the gag or env ~.~rot=eins of
Feline Immunodeficiency Viru~~ ~:FIV) :~r immunogenic
fragments thereof .. Crag anct :~ncT genes usef~.il in practicing
the present. invention may k~e ox~taine:_1 by mf~thods


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
4
well-known in the art. In one embodiment, viral RNA is reverse-transcribed
using
endogenous or exogenous reverse transcriptase and the DNA is rendered double-
stranded
using DNA polymerise. The gag and env-encoding DNA segments are then recovered
by
restriction enzyme digestion and are amplified by cloning in E. coli. In
another
embodiment, FIV-infected cat cells serve as a source of FIV proviral DNA. In
this
embodiment, chromosomal DNA is isolated from the cells, and oligonucleotide
primers
are used to specifically amplify the gag and env genes or fragments therefrom
using
polymerise chain reaction techniques. This approach is broadly applicable to
purifying
gag and env genes from different FIV strains or isolates, since primers can be
designed
from non-polymorphic regions of the FIV genome.
FIV gag and env genes isolated by the above methods are first inserted into
a transfer plasmid, and the recombinant plasmid is introduced into appropriate
host cells
that have been previously infected with a raccoon poxvirus. As a result, the
DNA from
the transfer plasmid is incorporated into the poxvirus DNA by homologous
recombination,
producing the RRPVs that are released from the cells.
DNA encoding the FIV gag or env proteins or fragments therefrom are
inserted into a transfer plasmid downstream of a poxvirus promoter. In a
preferred
embodiment, the early/late 7.5 kD protein promoter of vaccinia virus is used.
However,
alternate promoter elements could be used.
The preferred transfer plasmid also contains a beta-galactosidase marker
gene, which allows for selection and detection of the plasmid DNA sequences in
recombinant viruses. It will be understood by those of ordinary skill in the
art that
alternate selectable marker genes, such as the neomycin resistance gene or the
E. coli gpr
gene or others, could be used to practice the invention. Flanking the inserted
FIV gene
and the selectable marker gene are thymidine kinase DNA sequences, which
facilitate
integration of the plasmid DNA sequences into the raccoon poxvirus DNA by
homologous
recombination.
Recombinant viruses expressing the FIV gag or env genes are prepared by
first infecting a susceptible cell line (such as Vero [ATCC CCL 81], BSC-1
[ATCC CCL
26], RAT-2 [ATCC CRL 1764], or CRFK [ATCC CCL 94]) with wild type raccoon
poxvirus (ATCC VR-838 or similar isolates). Transfer plasmid DNA containing
the FIV


CA 02224257 1997-12-OS
WO 96/40268 PCTIUS96/08508
gag or env gene is then transfected into the infected cells using cationic
liposome-mediated
transfection, or other suitable techniques such as electroporation or calcium-
phosphate
precipitation. Raccoon poxviruses incorporate DNA from the transfer plasmid
through
homologous recombination with the thymidine kinase gene sequences present on
the
plasmid. Virus infection is allowed to proceed until cytopathic effects are
noted in all
cells.
Incorporation of the FIV gag or env gene into poxvirus DNA is
accompanied by disruption of the viral thymidine kinase gene. Thus,
recombinant virus
may be selected for by the absence of a thymidine kinase gene; this is
achieved by
selective expansion on RAT-2 cells (tk-, ATCC CRL 1764) in the presence of 5-
bromodeoxyuridine. Viruses containing a gene insert from the transfer plasmid
are
identified by blue plaque color when grown in the presence of a chromogenic
substrate for
beta-galactosidase such as X-gal.
Viral plaques that survive these selection and screening procedures are then
subjected to several cycles of plaque purification. Subsequently, the presence
of the gag
or env genes is confirmed by polymerise chain reaction technology, and the
presence of
gag or env antigenic determinant is confirmed by immunoblot analysis using
specific
antibodies. These viruses are designated by RRPV-FIV gag and RRPV-FIV env,
respectively.
In a further embodiment of the present invention, RRPVs can be produced
that express less-than-full-length segments of the FIV gag and env proteins.
The
techniques used to engineer transfer plasmids encoding partial sequences of
env and gag
are well-known ind widely used in the art, as are the methods for production
and
screening of RRPVs as detailed in this specification. For example, convenient
restriction
enzyme recognition sites can be used to obtain fragments of either gene, as
described,
e.g., Example 1 below. Alternatively, introduction of oligonucleotides
containing a stop
colon at various points along gag or env DNA will produce a nested set of
carboxyterminal-truncated versions of that gene, which can then be
incorporated into
RRPVs. Furthermore, sequences that encode different domains on each protein
may be
' recombined, using domains derived from different ~ FIV strains or isolates.
It will be
apparent to one of ordinary skill in the art that systematic screening of such
recombinant


CA 02224257 1997-12-OS
WO 96/40268 PCTIUS96/08508
6
RRPVs can establish whether the intact protein, subfragments thereof or mufti-
strain
recombinants thereof, are most preferred in practicing the present invention.
Furthermore, as stated above, DNA encoding different fragments of gag and env
can be
used in a combination vaccine after incorporation into the same, or different,
RRPVs.
For vaccine preparation, susceptible cells are grown in minimum essential
media containing fetal bovine serum or a suitable media substitute. Cells are
infected with
recombinant raccoon poxvirus at a multiplicity of infection of 0.1 infectious
units/cell or
less. In this specification an infectious unit is defined as a Tissue Culture
Infectious Dose
(TCID~), an amount of virus yielding 50% infection under defined conditions.
When
cytopathology is noted in > 90 % of the cells, the infected cells and
extracellular fluids are
harvested. The virus may be stored frozen (-50°C or colder) or
lyophilized until the time
of use. Compounds such as NZ-amine, dextrose, gelatin or others designed to
stabilize
the virus during freezing and lyophilization may be added. The virus may be
concentrated
using commercially available equipment.
Typically, the concentration of virus in the vaccine formulation will be a
minimum of 106'5 TCID~ per dose , but will typically be in the range of
10T° to 109'°
TCID~ per dose. At the time of vaccination, the virus is thawed (if frozen) or
reconstituted (if lyophilized) with a physiologically-acceptable carrier such
as deionized
water, saline, phosphate buffered saline, or the like.
In one embodiment, a physiologically acceptable adjuvant such as, for
example, EMA31, Adjuvant A, or combinations thereof, is added to the vaccine
formulation. Non-limiting examples of suitable adjuvants include squalane and
squalene
(or other oils of animal origin); block copolymers such as Pluronic~ (L121)
Saponin;
detergents such as Tween~-80; Quil~ A, mineral oils such as Drakeol~ or
Marcol~,
vegetable oils such as peanut oil; Corynebacterium-derived adjuvants such as
corynebacterium parvum; Propionibacterium~erived adjuvants such as
Propionibacterium
acne; Mycobacterium bovis (Bacillus Cahnette and Guerinn, or BCG);
interleukins such as
interleukin 2 and interleukin-12; monokines such as interleukin 1; tumor
necrosis factor;
interferons such as gamma interferon; combinations such as saponin-aluminum
hydroxide
or Quil~-A aluminum hydroxide; liposomes; iscom adjuvant; mycobacterial cell
wall
extract; synthetic glycopeptides such as muramyl dipeptides or other
derivatives; Avridine;


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
7
Lipid A; dextran sulfate; DEAF-Dextran or DEAF-Dextran with aluminum
phosphate;
carboxypolymethylene, such as Carbopol~; EMA; acrylic copolymer emulsions such
as
Neocryl~ A640 (e.g. U.S. Patent 5,047,238); vaccinia or animal poxvirus
proteins;
subviral particle adjuvants such as orbivirus; cholera toxin;
dimethyldiocledecylammonium
bromide; or mixtures thereof.
EMA 31 (Monsanto, St. Louis, MO) is a linear ethylene/maleic copolymer
with approximately equal amounts of ethylene and malefic anhydride, having an
estimated
average molecular weight of about 75,000 to 100,000. Adjuvant A is an adjuvant
comprising a block copolymer, such as a polyoxypropylene-polyoxyethylene (POP-
POE)
block copolymer, preferably Pluronic~ L121 (e.g. U.S. Patent 4,772,466), and
an organic
component, such as a metabolizable oil, e.g. an unsaturated turpin
hydrocarbon,
preferably squalane (2,6,10,15,19,23-hexamethyltetracosane) or squalene. The
vaccine
may also ilxlude a non-ionic detergent or surfactant, preferably a
polyoxyethylene sorbitan
monooleate such as a Tween~ detergent, most preferably Tween~-80, i.e.
polyoxyethylene
(20) sorbitan monooleate.
In this adjuvant mixture, the block copolymer, organic oil, and surfactant
may be present in amounts ranging from about 10 to about 40 mI/L, about 20 to
about 80
mI/L, and about 1.5 to about 6.5 mI/L, respectively. In a preferred embodiment
of the
stock adjuvant, the organic component is squalane present in an amount of
about 40 mL/L,
the surfactant is polyoxyethylenesorbitan monooleate (Tween~-80) present in an
amount of
about 3.2 mIIL, and the POP-POE block copolymer is Pluronic~ L121 present in
an
amount of about 20 mI/L. Pluronic~ L121 is a liquid copolymer at 15-
40°C, where the
polyoxypropylene (POP) compo~nt has a molecular weight of 3250 to 4000 and the
polyoxyethylene (POE) component comprises about 10-20 % , preferably 10 % , of
the total
molecule.
Individual raccoon poxviruses expressing the gag or env genes may be
mixed together for vaccination. Furthermore, the virus may be mixed with
additional
inactivated or attenuated viruses, bacteria, or fungi, or with immunogens
derived from
viruses, bacteria, or fungi such as feline leukemia virus, feline
panleukopenia virus, feline
rhinotracheitis virus, feline calicivirus, feline infectious peritonitis
virus, feline Chlamydia
psittaci, Microsporum canis, or others. In addition, antigens from the above-
cited


CA 02224257 1997-12-OS
WO 96/40268 . PCT/US96/08508
8
organisms may be incorporated into combination vaccines. 'These antigens may
be
purified from natural sources or from recombinant expression systems, or may
comprise
individual subunits of the antigen or synthetic peptides derived therefrom.
In a further embodiment of the present invention, live or inactivated RRPV
virus-cell lysates can be incorporated into liposomes, or encapsulated in
peptide-, protein-,
or polysaccharide-based microcapsules prior to administration, using means
that are
known in the art. The vaccine of the present invention is administered to cats
in volumes
ranging from 0.5 to 5 milliliters. The vaccine can be administered to cats by
subcutaneous, intramuscular, oral, intradermal, or intranasal routes. The
number of
injections and their temporal spacing may be varied. One to three vaccinations
administered at intervals of one to three weeks are usually effective.
The efficacy of the vaccines of the present invention is assessed by the
following methods. At about one month after the final vaccination, vaccinates
and
controls are each challenged with 3-20 cat ID~ units, preferably 5 cat ID~
units of FIV,
preferably the NCSU1 isolate (ATCC VR-2333). Whole blood is obtained from the
animals immediately before challenge, and at intervals after challenge, for
measurement of
a) viremia and b) relative amounts of CD4 and CD8 lymphocytes.
Viremia is measured by isolating mononuclear cells from the blood, and co-
culturing the cells with mononuclear cells from uninfected animals. After 7
days of
culture, the culture supernatants are tested for FIV by enzyme-linked
immunoassay (See
Example 5 below).
The ratio of CD4 to CD8 lymphocytes in the circulation of vaccinates and
controls is taken as a measure of immune function. Typically, FIV infection
causes an
inversion of the normal CD4:CD8 ratio of about 1.5-4.0 to a pathological ratio
of about
0.5-1Ø The numbers of CD4 and CD8 lymphocytes are measured by flow cytometry
using specific antibodies (see Example 5 below).
Another measure of immune function is to challenge vaccinates and
controls with Toxoplasma gondii at 6-12 months after the final RRPV-F1V
vaccination.
Normally, the severity of T. gondii- induced disease symptoms is considerably
exacerbated
in FIV-infected cats relative to uninfected cats. The severity of the T.
gondii effect is
determined by scoring ocular discharge, nasal discharge, dyspnea, and fever.


CA 02224257 1997-12-OS
WO 96/40268 PCTIUS96/08508
9
It will be understood that amelioration of any of the symptoms of FIV
infection is a desirable clinical goal. This includes a lessening of the
dosage of medication
used to treat FIV-induced symptoms.
The following examples are intended to illustrate the present invention
. without limitation thereof.
A. Isolation of Viral DNA
FIV strain NCSU-1 (designated "FIV-NCSU-1 ") was isolated from a
naturally infected, feline leukemia virus-negative cat and has been described
previously
(Tompkins et al., J. Am. Vet. Med. Assoc. 199: 1311, 1991. The virus was
passed in a
normal specific pathogen-free (SPF) cat (obtained from Liberty Laboratories,
Waverly,
N~. FIV-infected peripheral blood mononuclear cells (PBMC) were obtained from
whole blood by separation on discontinuous percoll gradients. Briefly, anti-
coagulated
whole blood was layered over a two step gradient containing 43 % PercollTM
(Pharmacia,
Piscataway, Nn over 62.5 % PercollTM in 0.15 M NaCI. Gradients were
centrifuged at 400
x g for 5 minutes, followed by 800 x g for 20 minutes at 22°C. PBMC
were harvested
from the gradient interface and washed in phosphate buffered saline containing
5 % fetal
bovine serum. In parallel, PBMCs were isolated from normal cats.
FIV was propagated by co~ulture of PBMCs from an FIV-infected cat with
PBMCs from normal cats. The cells were maintained in RPMI 1640 media
containing
% fetal bovine serum, 2.5 x 10-5 beta-mercaptoethanol, 2 mM L-glutalnine, 5
p,g/mL
concanavalin A, and 20% conditioned media from MLA cells (ATCC TIB 201) as a
source of interleukin-2 (II,-2).
Cat genomic DNA containing FIV-NCSU-1 proviral sequences was isolated
from the cultured PBMCs by lysis of the cells with 0.6 % sodium dodecyl
sulfate (SDS) in
10 mM Tris-HCI, pH 7.5, 10 mM EDTA, followed by precipitation of chromosomal
. DNA by incubation overnight with 1 mM NaCI. The DNA was recovered by
centrifugation at 10,000 r.p.m. (Beckman J2, JA-20 rotor) for 40 minutes. The
DNA
' pellet was resuspended in a solution containing 10 mM Tris-HCl pH 7.5, 10 mM
EDTA,
0.1 % SDS buffer and digested with ribonuclease A (20 p.glml) and proteinase K
(0.2


CA 02224257 2003-02-19
WO 96/40268 PCTNS96/085J8
mghnl) at 50°C for 4 hours. DNA was then purified by sequential
extraction with phenol,
phenol:chlomfortn (1:1) and chloroform, and was recovered in pure form
followed by
ethanol precipitation.
CloninE o~ Envelope Gene
FIV-NCSU-1 envelope DNA sequences were cloned using polymerise
chain reaction (PCR) methods as follows:
~~nvelooe Fragment A
The following oligonucleotides were used to amplify the 5' proximal
segment of the env gene.
5'-TCGGATSCA-ACAATAATT"ATGGCAGA_AG~-3' [SEQ. LD. NO. 1] (Coding
strand, 6252-V)
5'-AATCAGGTACAAAGTCACCGTTC-3' [SEQ. LD. NO. 2] (Complementary strand,
6745-C)
Primer 6252-V cormsponds to nucleotides 6252-6273 of FIV strain PPR
(GenBank No. M36968) and primer 6745-C (underlined region) corresponds to
nucleotides
6723-6745 of FIY strain 14 (GenF~ank No. 25381). The start colon for envelope
protein
translation is included in primer 6252-V. Primer 6252-V also has a synthetic
BamHI
restriction enzyme site near the 5' end to facilitate cloning. An AvrII site
located at position
6719 also facilitates cloning. Envelope fragment A is 494 by in length.
nv
The following oligonucleotides were used to amplify the middle segment of
the tnv gene.
5'-TATAGAAGCACCCCAAGAAGAG-3' [SEQ. LD. NO. 3] (Coding strand, 6637-V)
5'-CATTCCCCCAAAGTTATATTTC-3' [SEQ. LD. NO. 4] (Complementary strand,
8469-C)
Primers 6637-V and 8469-C correspond to nucleotides 6637-6659 and
8448-8469 of FIV 14 strain, respectively. An Avrll site at position 6719 and a
Spec site
at position 8288 facilitated cloning. Envelope fragment B is 1833 by in
length.
"'T'rade-mark

CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
11
The following oligonucleotides were used to amplify the 3' distal fragment
of the env gene.
5'- TTAGTTACATTAGAGCATCAAG-3' [SEQ. LD. NO. 5] (Coding strand, 8264-V)
5'-TTCTAGATCTTCAC~'~('T ATA T -3' [SEQ. LD. NO. 6] (Complementary
strand, 9145-C)
Primer 8264-V corresponds to nucleotides 8264-8285 of FIV strain 14, and
primer 9145-C (underlined region) corresponds to nucleotides 9126-9145 of FIV
strain
PPR. Primer 9145-C has a synthetic BgIII site near the 5' end to facilitate
cloning. An
SpeI site located at position 8288 also facilitated cloning. Envelope fragment
C is 880 by
in length.
In each case, PCR was performed for 35 cycles of 1 min 30 sec at
94°C, 2
min at 56°C, and 2 min at 72°C, followed by one cycle of 8 min
at 72°C. Each envelope
fragment was isolated by gel electrophoresis and cloned into plasmid pSL1190
using
standard methods (Maniatis et al., Molecular Cloning: A Laboratory Manual,
1982, Cold
Spring Harbor Press).
Initially, each fragment was cloned into pSL1190, after which the three
fragments were spliced together to recreate a full length envelope gene. For
this purpose,
the Envelope A plasmid was digested with BamHI and AvrII, the envelope B
plasmid was
digested was with AvrII and SpeI, and the envelope C plasmid was digested with
SpeI and
BgIII. Subsequently, the 1.5 kbp AvrII/SpeI envelope B fragment was ligated
into pSL-
EnvA that had been digested with AvrII and SpeI to create pSL-EnvAB (Figure
1). The
envAB fragment codes for the entire surface membrane protein (SU) and the
first 63
amino acids from the amino-terminus of the transmembrane protein (TIVI) of FIV-
NCSU-
1, i.e., amino acids 1 - 735 of env. However, envAB does not contain the
transmembrane
domain (T11~.
Next, the 0.9 kbp SpeIISmaI envelope C fragment from pSL-EnvC was
ligated into pSL-EnvAB that had been digested with SpeI and BbrPI, to create
pSL-
EnvABC or pSL-WEnv (Figure 1). The WEnv fragment codes for the entire env open
reading frame (SU a~ TM proteins) of FIV NCSU-1 (Figure 2).


CA 02224257 2003-02-19
-12
The subclonec:I clenE,t~:ic: el.ernents of FIV-NCSU-1 wer~a
sequenced using Sequenase (trade-mark) Version 2.0 (United
States Bi_ochemicaJ_, CleVel.anc.l, OH) as c~tesr.r_ ibed for double-
st.randed DNA, and t he reac:ticms were analyzed using t.ne ABI
automated sequences (Applied Biosystems, f~~:~ster City, CA).
Both D1'fA strands were sequen~::e..-i to confirrn the results .
The DNA sequences were analyze~_1 using the MacVector DNA
Analysis software (International Biotechno.logies, Inc., New
Haven, CT) . The e:~r~.',~ DNA segue~ces were analy:~ed for open
reading frames anccompared to the previously published DNA
sequenc;e~> of othea- fIV isolates. 'The DNA ;end predicted
amino acid sequenc.:es of env and envAB cpen reading frames
of FIV-Nf,SU-7_ are shown in figure 3.
C. Cloning of The FIV GAG Gene
The gag gene c;~f: F7=V-NCS'~:1: was arnplifie:d using PCF; and
the fol.lowin~~ olic;onucleotid~.~ primers
5' -CAATTCTAGAGAGA('TCT.A~:AGCAA.<~ATG-3' [ SEQ. L . D _ NO. 7 ]
(codincJ strand, 6 ~,C;__v~)
5'-TAAfAGATCTGGCC~.'CTTTTCTAATGATG-3' [SEQ. I. D. NO. 8]
(Complementa:ry str_a.nd, ?026-C:)
Primers 610-~% and % 026-<' :vorresr~ond t:~o nucleotide=s
610-630 and ?005-~_'0?6 of FIV 14 strain, respectively.
Primer; 610-V arid 2026--C have: ;x:baI and Bg.iLI restriction
enzyme sites, res~:~ecti~~ely, near their_ 5' ends to
facilit.at:e c.l_oninc~. The last: three nucleotides of primer
610-V correspond t:o the st.ar~f: :o~~on for gag protein
translation. PCR was performed for 35 cycles of 1 min 30
sec at 94 ° C, 2 mir7 at 56 ° C, ~:rnc1 2 min at J;:' C,
followed by
one cycle of 4 min at ~2~C. The 1.4 },bp DNA fragment
containing the ga<gene was purified by ge;.L electrophoresis
and cloned into tree XbaI/Bgl:LI site of pSh:L190 to form pSL-
WGag (Figure 4) . The DNA seyluence of FIV-NCSU-1 gag is
shown i.n Figure 5.


CA 02224257 2003-02-19
-13-
Example 2: Preparation of Recombinant Raccoon Poxviruses
A. Construction of Raccoon Poxvirus Transfer Plasmids
The DIJA segue=~r:~r..ES enc;odin,-t the gag, E~nv, and env.~lB
isolated as described in Example 1 were in~:~ividually
subcloned into the I:>oxvirus t: ransfer vector p SC11 . T:ne
sequence of pSCl1 is~ discl.os~~d in co-pending Canadian
patent application Serial Nc. 2,132,3'74 laid open Mar~~h 23,
1995. For this purpose, the 1.4 kb XbaI/BglII: fragme:zt of
pSL-WGag (Figure F:), the 2.9 kb BamHl/SpiT fragment of pSL-
WEnv (figure 7) arid t:he :?.1 ~:b BamHI/.~pel: .fragment of pSL-
EnvAB lFi_gure 8) urere indi.vir_lually isolate~ci and rendered
blunt-ended with t=he Klenow fragment of DNA polymerase,
after Hrhich each ~,as individually cloned a.nto the SmaI site
of pSCl.l .
B, Preparation of Recombinant Raccoon Poxviruses
Recombinant raccoon pox v:i.ruses (R.RPV? bearing the FIV
gag and env genes were prepared as generally described
for recombinant va~~cinia vira.Ases (Mackett <:~nd Smith, J Gen
Virol Ei7: 206'; -2084:, 1.986 ) wi t h some modif:ir:ati.ons .
Mono layers of Verc cells (ATC:C CCL 81) that. were 80',
;:onfluent (approxi.mately 5 x 10" ~~e11_s in 1.00 mm tissue
culture: :fishes) wF~re. infecte~:l ~,aith wild-type raccoon pox
virus (ATCC VR-838) at a multiplicity of infection (MOI) of
0.1 TCIDS /cell in .... nvl of ME:M (Eagle's Mi.nimurn Essential
Medium (~lrJCO BRL #410-1500) containing 0. t)5','~ lactalbumin
hydrolysate and 15 ~g/ml gen:amicin sulfat~s~, adjusted to pH
'7.2 with sodium ba.carbonate) f.r.,r 30-60 minutes at 37 C.
The cells were then transfec'-ed with either pSCl1-FIV gag,
pSCl1-fIV env, or pSCll.-FIV c~nv AB t:ransf:e:r_ p-~asmids by
cationic liposome--mediated t:raosfect.ion using Transfectam~k:~
(Promega Cortporati.on, Madison, Wisconsin) according to
manufacaurer's in;tz:u;st:ions. 'Phe cells/L)N.t'~-liposomes


CA 02224257 2003-02-19
~- 13a -
mixturE: was incubat:ec~ in 3 rr:i_ of MEM cont:wining 5a, fetal
bovine serum ( FBS; oVer_night at 37 ' C ( 5''s C~:O_ ) , after which
the medium was red-~7..ac:ed with 8 ml fresh MF~M/5'': FBS. The
transfected ~~~ells were incubated at 37-'C (:p'<; ("O~) until
greater: than 80'a; c:~fthe cells showed c;ytopathic effects
(appro~:imate7_y 3-4 days) . T':ne cells and <:aiture media
(viral-cell )_ysate-s) were then removed from the plates and
subject: t:o two cy<::.les of fre~~zc,-thawing be:~ore storage at -
70'C.
C. Isolation of Recombinant Raccoon Pox Virus
Carrying the FIV gag Gene
RRPV carr:yinc~ the E'IV-NCSU,g_rg gene (RRPV-F~'IV gag) are
isolated and purii_ieci from t_re pSCl1-FIV c~,ag/Vero cel.1
transfect:ion by st:andat:d vir.-~l plaque assay methods.
Monolayers of Verc:~ cells (50-8~~'<'~ confluent.; in 100 mm
tissue cultui°e di;:lu.es were


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
14
infected with 2 ml of.l0-fold serial dilutions (10-' to 10-3 in MEM) of the
viral-cell lysates.
After incubation, for 1 hour at 37°C, the media are removed and the
infected cells were
overlaid with 8-10 ml of 1.25 % Noble agar containing MEM/S % FBS. The
infected cells
were then incubated for 3-4 days at 37°C (5 % COZ), and overlaid again
with 4 ml of
1.25 % Nobel agar containing O.SX PBS and 600 ug/ml S-bromo~-chloro-3-indolyl-
~3-D-
galactopyranoside (X-gal, United States Biochemical, Cleveland, Ohio). The
plates were
incubated at 37°C (5 % C02) for 4-16 hours, until blue viral plaques
((3-galactosidase
positive) were observed. The recombinant viral plaques were picked with
sterile blunt
needles attached to a 1 cc syringe, suspended in 0.5 ml of 0.25 mg/ml trypsin,
vortexed
vigorously, and incubated at 37°C for 15-30 minutes. The disrupted
viral plaques were
then inoculated onto 5x105 Vero cells in T-25 cm2 flasks and incubated at
37°C (5% C02)
until greater than 80% CPE was observed. The viral-cell lysates containing
RRpV_FIV
gag were subjected to two cycles of freeze-thawing and stored at -70°C.
Five individual
RRPV-FIV gagclones were selected and plagued purified four times as described
above.
D.
RRPV carrying the FIV-NCSU, envAB gene (RRPV-FIV envAB) were isolated and
purified from the pSCll-FIV envABIVero cell transfection using the methods as
described
for RRPV-FIV gag with some slight modifications. Thymidine kinase deficient
(tk-)
raccoon pox viruses from the initial viral~ell lysates were selected on tk-
Rat-2 cells
(ATCC CRL 1764). This was performed by inoculating 1 ml of the initial viral-
cell lysate
onto a monolayer of Rat-2 cells in a T-75 cm2 flask (approximately 5x106
cells) containing
S-bromodeoxyuridine (BrdU) at 30 ug/ml in MEM. The infected monolayer was
incubated at 37°C (5 % CO~ for 3-4 days until greater than 70 % CPE was
observed. The
tk- viral-cell lysates were subjected to two cycles of freeze-thawing two
times and stored at
-70°C. RRPV-FIV envAB were isolated and purified from the tk- viral-
cell lysates by the
standard viral plaque assay as described above for RRPV-FIV gag on Vero cells.
Five
individual RRPV-FIV env AB clones were selected and plaque purified five
times.


CA 02224257 1997-12-05
WO 96/40268 PCT/US96/08508
Example 3: Characteristics of Recombinant FIV-Expressing Raccoon Pox Viruses
A. Confirmation of FIV gaE and evnAB Genes in RRPV by Polymerise
Chain Reaction
The presence of the FIV gag and envAB genes in the RRPVs was
confirmed using PCR. 90 ul of a viral-cell lysate was incubated with 10 ~1 of
lOX PCR
lysis buffer (1X; 10 mM Tris-HCI buffer, pH 8.5, containing 50 mM KCI, 2.5 mM
MgClz, 0.5 °& Tween 20, 0.3 mglml Proteinase K) for 16 hours at
50°C, then boiled for
10 minutes. 10 ~cl of this lysate was used as a template in the PCR reaction.
PCR was
performed in 100 ~cl of 10 mM Tris-HCl buffer, pH 8.3, containing 50 mM KCI,
200 uM
of each dNTP, 1.5 mM MgCh, 30 pmoles of each primer, and 2.5 Units of
AmpliTaq~
DNA polymerise (Perldn-Elmer Cetus, Norwallc, CT). The primers used in the PCR
for
FIV gag were:
5'-TATGGAAAAGGCAAGAGAAGGAC-3'[SEQ. LD. NO. 9]
5'-TCGAGATACCATGCTCTACACTG-3', [SEQ. LD. NO. 10]
conresponding to nucleotides 471-493 and 763-785 of the FIV gag open reading
frame,
respectively. The primers used in the PCR for FIV envAB were:
5'-TATGGAAAAGATGGGATGAGACTA-3' [SEQ. LD. NO. 15]
5'-GTCACTTACCTTCATAGTAAACC-3' [SEQ. LD. NO. 16]
corresponding to nucleotides 857-880 and 1513-1535 of the FIV env open reading
frame,
respectively. The PCR amplifications were performed in a DNA Thermal Cycler
(Perkin-Elmer Cetus) by first heating the reaction mixes to 94°C for
denaturation, and
then 35 cycles of 1 minute at 95°C, 1 minute at 55°C, and 2
minutes at 72°C, and a final
incubation of 8 minutes at 72°C. 10 ~cl of the PCR products were
analyzed by
electrophoresis in a horizontal-submarine 4 % NuSieve~ agarose (FMC
BioProducts,
Rocldand, ME) gel in TAE buffer (40 mM Tris base, 20 mM sodium acetate, 1 mM
EDTA, pH ?.2) by applying 5 Vlcm for 1-2 hours, and staining with ethidium
bromide.
PCR amplifications with the FIV gag and env primers gave expected DNA
fragments of
314 and 678 nucleotides, respectively. PCR amplifications using the pSCll FIV
gag and
envAB transfer plasmids served as positive controls. PCR amplifications using
wild-type
raccoon pox virus-Vero cell lysates served as a negative control.
SUBSTITUTE SHEET (RULE 26)


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
16
B.
Confluent monolayers of Vero cells in a T-25 cm2 flask (1-2 x 106 cells)
were infected with clones of either RRPV-FIV gag or RRPV-FIV envAB at an
M.O.I. of
1 to 10 TCID~ per cell. The infected cells were incubated at 37°C (5 %
C02) for 2-3 days
until approximately 80% CPE was observed. 20 pl of the viral-cell lysate was
added to 5
p,l of SX Laemmli sample buffer (0.3 M Tris-HCl buffer, pH 6.8, containing 5 %
SDS,
50% glycerol, 0.4% bromophenol blue, and 3% 2-mercaptoethanol) and heated at
95°C
for 5 minutes. The denatured protein samples were separated by
SDS/polyacrylamide
electrophoresis using a 4-15% gradient polyacrylamide gel (Maniatis et al.,
Molecular
Cloning: A Laboratory Manual, 1982, Cold Spring Harbor Press). After
electrophoresis,
the proteins were transferred to nitrocellulose filters (Bio-Rad Laboratories,
Hercules, CA)
by electrotransfer using a Bio-Rad transfer apparatus per manufacturer's
instructions. The
transfer was performed in 25 mM Tris-HCl buffer, containing 0.2 M glycine and
20
methanol, for 45 minutes at SOV with constant current.
The blot was then screened for FTV gag and envAB proteins by
immunoblot analysis as previously described (Davis et al., Basic Methods in
Molecular
Biology, 1986, Elsevier Science Publishing Company, New York, NY) with some
slight
modifications. After transfer, the nitrocellulose blot was rinsed in phosphate
buffer
saline, pH 7.4, containing 0.1 % Tween-20 (PBS-TV~, and non-specific sites
were blocked
by incubating the blot in PBS containing 1 % bovine serum albumin (PBS-BSA) at
4°C
overnight, followed by a 15 mirnite wash in PBS-TW. The blot was then
incubated for 30
minutes at room temperature with goat anti-FIV IgG diluted 1:100 in PBS-TW
containing
1 % BSA (PBS-TW-BSA), followed by four 5 minute washes in PBS-TW. Next, the
blot
was incubated for 30 minutes at room temperature with a biotin-labeled mouse-
anti-goat
IgG antibody (secondary antibody) (Kirkegaard & Perry Laboratories Inc.,
Gaithersburg,
MD) diluted 1:2000 in PBS-TW-BSA, followed by four 5-minute washes in PBS-TW.
Antigen-antibody complexes were detected by, incubating the blot for 30
minutes at room
temperature with horseradish peroxidase-conjugated streptavidin (Kirkegaard &
Perry
Laboratories Inc.) diluted 1:1000 in PBS-TW, washing four times for 5 minutes
each in
PBS-TW, and visualizing with peroxidase chromogenic substrate (Kirkegaard &
Perry


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
17
Laboratories Inc.). Sucrose-gradient purified FIV and wild-type raccoon pox
virus/Vero
cell lysates were used as the positive and negative controls for the
immunoblot analysis,
respectively.
Goat anti-FIV antibodies were prepared as follows. FIV NCSU1 was
grown in peripheral blood lymphocytes and concentrated using a hollow fiber
apparatus to
a concentration of about 106 TC>Ds~/ml. The concentrated virus stock was mixed
with an
oil adjuvant such as OW3 in a ratio of 1:1 (v: v), and the emulsion was used
to inoculate
goats six times, at intervals of 3-4 weeks. At monthly intervals, the goats
were bled and
the serum was tested for the presence of anti-FIV antibodies.
C. Confirmation of IZRPV FIV ~aø and nv R pro Pin F~n~r ~;.,n ~,s
ImmLn_oflLOrescence cav
Confluent monolayers of Vero cells in 96-well plates (1-2 x 104 cellslwell)
are infected with clones of either RRPV-FIV gag or RRPV-FIV envAB at a
multiplicity of
infection of 0.1 to 1.0 plaque forming units per cell. Cell infected with wild-
type RCNV
serve as a negative control. The infected cells are incubated at 37°C
(5 % C02) for 1 day
until approximately 20% CPE is observed. The cells are then washed three times
with
PBS and fixed with 80% acetone at 4°C for ten minutes. Next, the cells
are rehydrated
with PBS and incubated with a monoclonal antibody (IgG) against either FIV gag
or env
surface membrane proteins for 30 minutes at room temperature. FIV antigen/FIV
antibody complexes are detected using a FTTC-conjugated goat anti-mouse IgG
(Kirkegaard & Perry Laboratories Inc., Gaithersburg, MD) and fluorescence
microscopy.
D.
Virus preparations were pre-treated by dilution into an equal volume of
0.5 % trypsin and incubation at 37°C for 30 minutes in order to release
virus from
inclusions. Serial dilutions (10-fold) of virus were then prepared in MEM and
were
inoculated (100 p,l/well) in replicates of five onto Vero cells (1 x 104 cells
in 100 pl per
well) in a 96 well plate. Plates were incubated for 3-5 days at 37°C (5
% C02) and
observed for cytopathology typical of raccoon poxvirus. Viral infectivity
titers were
calculated as 50% endpoints based on cytopathology using the methods of Reed
and
Muench (Reed and Muench, The American Journal of Hygiene ZZ: 493, 1938).


CA 02224257 1997-12-OS
WO 96/40268 . PCT/US96/08508
18
Exampile 4: Pren~ration of Vaccines Bas~d on Recombinant Pn~ m;...,~o~
A.
A single clone of each recombinant virus that showed significant
recombinant protein expression (by the method described in Example 3 above)
was
selected for large-scale expansion to serve as a master seed virus. All
recombinant virus
expansions and titrations were done on Vero cells in MEM containing 2.5 % FBS.
Each
plaque-purified virus clone was expanded by inoculating a confluent monolayer
of Vero
cells in a T-150 cm2 flask (1x10' cells) with 1 ml of viral-cell lysate
(approximately 10'
infectious virus particles), and incubating at 37°C (5 % C02) until 100
% CPE was
observed (2-3 days). , Thin viral-cell lysate served as a pre-master seed
virus stock and was
used to obtain the master seed virus. The pre-master seed of each recombinant
virus was
titrated on Vero cells and a TCID~ was determined. The master seed viruses for
RRPV-
FTV gag and RRPV-FTV envAB were grown on Vero cells using an MOI of 0.01 and
0.1,
respectively. Three roller bottles of confluent Vero cells were infected for
each of the
master seed viruses using MEM media supplemented with 2.5 % fetal bovine serum
and
incubated for approximately 3 days at 37°C. Infected culture
supernatant fluids were
harvested, and seed viruses were aliquoted into 1.5 ml ampules, which were
sealed and
stored in a liquid nitrogen freezer.
B. Preparation of Vac inPc
Vero cells (3 x 10') were seeded into 850 cm2 roller bottles in 200 ml of
growth media (MEM containing 0. 5 % lactalbumin hydrolysate and 5 % heat-
inactivated
fetal bovine serum) and incubated for 18 hours at 37°C. The next day,
the media were
removed from the cells and replaced with 50 ml of RRPV-FN gag at a
multiplicity of
infection of 0.01 in infection media (MEM containing 0.5 % LAH and 2.5 % heat-
inactivated fetal bovine serum). The virus used was at the fourth passage
beyond the
master seed preparation. Virus was allowed to adsorb to the cells for 30
minutes at 37°C, '
after which the volume of medium was adjusted to 150 ml per roller bottle.
Roller bottles
were incubated at 37°C until 100% cytopathology was evident (3 days). A
viruslcell


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
19
lysate was then prepared and stored frozen (-70°C). The virus titer of
RRPV-FIV gag was
determined to be 10''4 TCID~/ml.
Recombinant raccoon poxviruses expressing the FIV envAB gene fragment
were prepared in the same manner, except that a multiplicity of infection of
0.1 was used.
The virus was at the fourth passage beyond the master seed preparation. The
RRPV-FIV
envAB preparation was titered and found to contain 1064 TCIDsdml.
Wild type raccoon poxvirus was grown using the same methods as
described above. This virus preparation was found to contain 10''' TCID~/ml.
A. Vaccination
Thirty 6-7 month old cats (specific pathogen-free, Harlan Sprague Dawley,
Madison, Wn, fifteen males and fifteen females, were used. Cats were divided
into six
groups and vaccinated twice, 21 days apart, as indicated below:
Table 1. Assignment of Groups for Vaccination
Group # Cats Vaccine Volume Virus Route


Dose(TCm~)


1 5 RRPV-FIV gag 1 mL 10''4 SC


2 5 RRPV-FIV gag 1 mL 10''4


3 5 RRPV-FIV envAB 3 mL 106'9 SC


4 5 RRPV-FN envAB 2 mL 106''


5 RRPV-FIV gag(1 ml)+4 mL 10''4 (gag) SC


RRPV-FTV envAB 3 mL 106'9 (envAB)SC


6 5 Wild Type raccoon pox 1 mL 10''' SC
virus
~ SC = suewrumus vaccimtion
IM = Intrartuucular Vxcimtion


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
20 -
B. ~~erimental DeciPn
Twenty-five cats were vaccinated with the recombinant raccoon poxvirus
vaccines as indicated in Table 1. Five cats were administered a similar titer
of wild type
raccoon poxvirus to serve a negative controls. Two vaccinations were
administered 21
days apart. Subcutaneous vaccinations were administered in the nape of the
neck, and
intramuscular vaccinations were administered in a rear thigh. Four weeks
following the
second vaccination, all cats were challenged with the NCSU-1 strain of FIV and
monitored
for viremia and evidence of lymphocyte population changes as described below.
Eleven
months following FIV challenge, cats in Groups 1, 2, 3, 4, and 6 were
challenged with
Toxoplasma gondii and monitored for 48 days for clinical signs of disease.
C.
Four weeks following the second vaccination, all of the cats were
challenged subcutaneously with 10 cat ID~ units of the NCSUI isolate of
FIV(1:1000
dilution of lot # 021891). Whole blood was obtained from the cats prior to
challenge, and
periodically after challenge, in order to assess virus infection parameters as
follows:
1. Detection of Viremia
Culture isolation of FI:V was performed as described previously (Wasmoen
et al., Vet. Immuno. Immrtnopath. x:83 1992). Mononuclear cells were isolated
from
whole blood using Percoll~ (Pharmacia Biotech, Piscataway NJ) gradients. 5 x
105 cells
from FIV-challenged cats were cultured with 1 x 106 mononuclear cells isolated
from
uninfected cats. Cultures were fed with RPMI media every 7 days and
supernatants tested
for the presence of FIV by an enzyme-linked immunosorbent assay (ELISA) that
detects
Fl'V p25 antigen (Petcheck ELISA, )DEXX, Portland ME).
2. j~y~hocvte ubce c
Leukocytes were isolated from whole blood using HistopaqueTM (Sigma
Chemical Company, St. Louis MO) and lymphocyte subsets quantitated by staining
the
cells with antibodies specific to CD4 (monoclonal antibody CAT30A), CD8
(monoclonal


CA 02224257 1997-12-OS
WO 96/40268 PCT/US96/08508
21
antibody FLSM 3.357), pan T lymphocytes (monoclonal antibody FLSM 1.572) or B
lymphocytes (anti-cat IgG) followed by FACS analysis. These monoclonal
antibodies are
described elsewhere (Tompkins et al. Vet. Irrrmunol. Irnmunopathol. xø:305,
1990) and the
flow cytometry procedure is the same as previously described (R. V. English et
al. J.
Infect. Dis. ~Q:543, 1994). CD4:CD8 ratios were calculated.
D. ToYo ~ ~,yon%~ii Ch~llen
Tacheozoites of the ME49 strain of T. gondii that were frozen in 10%
glycerol were inoculated intraperitoneally into Swiss mice (Charles Rivers
Laboratories)
and serially passed in mice according to published procedures (Davidson et
al., Am. J.
Pathol. x:1486, 1993). Tacheozoites harvested from peritoneal fluids of mice
were
enumerated using a hemacytometer. Cats were tranquilized using ketamine
hydrochloride
and inoculated with 50,000 fresh tachyzoites into the right common carotid
artery that had
been surgically isolated. Cats were monitored for clinical signs of disease,
including
ocular discharge, nasal discharge, dyspnea, fever, depression, and weight loss
for 3 days
prior to and 48 days following T. gondii inoculation.
Clinical signs follow T. gondii challenge were scored as follows:
Clinical Sign Score
Fever 103.0 to 1 point per day
103.9°F
104.0 to 2 points per day
104.9°F
>_105.0F 3 points per
day


(temperatures
were not scored
until ?I F
above baseline.
)


Depression/Lethargy1 point per day


Dehydration 2 points per day


Nasal Discharge1 point per day


Ocular Discharge1 point per day


Respiratory
Distress:


Tachypnea 2 points per day


Dyspnea 4 points per day




CA 02224257 1997-12-OS
WO 96/40268 PCT/US96108508
22
E. ~~!:S~
At one month following inoculation with the NCSU-1 strain of FIV, 60%
of the control cats were found to be viremic (Figure 9). Cats vaccinated with
RRPV-FIV
gag were all negative for FIV, 40 % of the cats vaccinated with RRPV-FIV envAB
were
virus positive, and 20% of the cats vaccinated with a combination of these two
viruses
were viremic (Figure 9). Therefore, the ability of the test vaccines to
prevent viremia at
this time point varied from 33% to 100% (Figure 10).
At three months after FIV challenge, 80% of the control cats were found to
be virus positive (Figure 9). Similarly, FIV could be isolated from peripheral
blood
mononuclear cells of nearly all vaccinated cats using this very sensitive
method (Figure 9).
With respect to immune status, 80% of the control cats showed evidence of
CD4:CD8 lymphocyte ratio inversions (i.e. ratios less than 1.0) at three
months (Figure
9). In contrast, only 30% of the RRPV-gag vaccinated cats had evidence of
significant
CD4:CD8 inversions, and the RRPV-FIV envAB vaccinates were similarly protected
from
this lymphocyte subset change (Figure 9). Cats vaccinated with a combination
of the two
recombinant viruses were not significantly different from the controls (i.e.
80% showed
CD4:CD8 inversions) at 3 months after challenge (Figure 9).
At 9 months after FIV challenge, 100% of the control cats were FIV
infected, and all showed CD4:CD8 inversions (Figure 9). A large percentage of
the
vaccinated cats were also shown to be viremic at this time point. However,
only 50% of
the RRPV-FIV gag vaccinates and 20% of the RRPV-FIV envAB vaccinates showed
evidence of CD4:CD8 inversions at this time point. Therefore, these two
vaccines showed
a significant ability to prevent the CD4: CD8 lymphocyte ratio changes
associated with FIV
infection even though the cats appeared to be viremic (Figure 10).
In order to determine whether CD4:CD8 lymphocyte subset inversions
signified a deterioration in the immune system of cats following FIV infection
(and,
conversely, that lack of inversion in vaccinates signified maintenance of
immune function),
vaccinated and control cats (from groups 1, 2, 3, 4, and 6) were challenged
with
Toxoplasma gondii. This parasite causes subclinical infections in normal cats,
but has
been reported to cause severe disease in cats that are immunocompromised due
to FIV


CA 02224257 1997-12-OS
WO 96/40268 PCTIUS96/08508
23
infection (Davidson et al., Am. J. Pathol. x:1486, 1993). Following T. gondii
challenge, control cats displayed ocular discharge, nasal discharge, dyspnea,
and fever.
The average total clinical score for control cats was 15.6 (Figure 11). By
comparison,
there was a 41 % reduction in clinical disease scores in RRPV-FIV gag
vaccinated cats,
related to reductions in clinical signs of ocular discharge and dyspnea
(Figure 11). The
clinical picture following T. gondii challenge was even less severe in
RRPV_FIV envAB
vaccinated cats. This group showed a 92% decrease in ocular signs, 75%
decrease in
nasal discharge, 73 % reduction in dyspnea, and 58 % decrease in overall
clinical scores
(Figure 11). Further, 80% of the control cats displayed weight loss in the
first 14 days
after challenge, compared to weight loss in only 44% of the RRPV-FIV gag
vaccinates
and 50% of the V-FN envAB vaccinates. Therefore, control cats were more
susceptible
to induction of disease by this opportunistic pathogen than vaccinated cats.
These data suggest that vaccination altered the progression of clinical
disease
caused by this virus (i.e. induction of immune suppression). This is indicated
by a lower
rate of CD4:CD8 inversions in vaccinated cats and by a decreased
susceptibility to
infection with the opportunistic pathogen T. gondii.

Representative Drawing

Sorry, the representative drawing for patent document number 2224257 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2005-08-16
(86) PCT Filing Date 1996-06-03
(87) PCT Publication Date 1996-12-19
(85) National Entry 1997-12-05
Examination Requested 2001-05-30
(45) Issued 2005-08-16
Deemed Expired 2013-06-04

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $300.00 1997-12-05
Maintenance Fee - Application - New Act 2 1998-06-03 $100.00 1998-04-06
Registration of a document - section 124 $100.00 1998-11-27
Maintenance Fee - Application - New Act 3 1999-06-03 $100.00 1999-04-14
Maintenance Fee - Application - New Act 4 2000-06-05 $100.00 2000-03-21
Maintenance Fee - Application - New Act 5 2001-06-04 $150.00 2001-04-05
Request for Examination $400.00 2001-05-30
Maintenance Fee - Application - New Act 6 2002-06-03 $150.00 2002-03-20
Registration of a document - section 124 $50.00 2002-07-09
Maintenance Fee - Application - New Act 7 2003-06-03 $150.00 2003-03-20
Maintenance Fee - Application - New Act 8 2004-06-03 $200.00 2004-03-18
Maintenance Fee - Application - New Act 9 2005-06-03 $200.00 2005-03-16
Final Fee $300.00 2005-05-31
Maintenance Fee - Patent - New Act 10 2006-06-05 $250.00 2006-05-08
Maintenance Fee - Patent - New Act 11 2007-06-04 $250.00 2007-05-07
Maintenance Fee - Patent - New Act 12 2008-06-03 $250.00 2008-05-07
Maintenance Fee - Patent - New Act 13 2009-06-03 $250.00 2009-05-07
Maintenance Fee - Patent - New Act 14 2010-06-03 $250.00 2010-05-07
Maintenance Fee - Patent - New Act 15 2011-06-03 $450.00 2011-05-18
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
WYETH
Past Owners on Record
AMERICAN HOME PRODUCTS CORPORATION
CHAVEZ, LLOYD GEORGE, JR.
CHU, HSIEN-JUE
WASMOEN, TERRI
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Cover Page 1998-04-02 1 54
Description 2003-02-19 25 1,119
Claims 2003-02-19 4 114
Description 1997-12-05 23 1,095
Abstract 1997-12-05 1 52
Claims 1997-12-05 4 113
Drawings 1997-12-05 18 544
Claims 2004-05-19 4 116
Cover Page 2005-08-02 1 40
Fees 2000-03-24 1 32
Assignment 1998-11-27 5 214
Assignment 1997-12-05 2 102
PCT 1997-12-05 14 443
Correspondence 1998-03-16 1 32
Prosecution-Amendment 2001-05-30 1 51
Assignment 2002-07-09 10 277
Prosecution-Amendment 2002-11-08 2 60
Prosecution-Amendment 2003-02-19 11 325
Prosecution-Amendment 2003-03-27 1 31
Fees 2003-03-20 1 35
Prosecution-Amendment 2003-03-28 1 37
Fees 2001-04-05 1 33
Fees 2002-03-20 1 32
Fees 1998-04-06 1 31
Fees 1999-04-14 1 27
Fees 2004-03-18 1 35
Prosecution-Amendment 2004-05-04 1 27
Prosecution-Amendment 2004-05-19 2 64
Fees 2005-03-16 1 30
Correspondence 2005-05-31 1 25